22
1 Kupfer-type Immunological Synapses in vivo: Raison D’être of SMAC Izaskun Mitxitorena, Elena Saavedra, Carlos Barcia, Ph.D. Department of Biochemistry and Molecular Biology, Institute of Neuroscience & School of Medicine, Universitat Autonoma de Barcelona, Lab M2-107, Bellaterra, Cerdanyola del Valles, Barcelona, Spain. Running Title: SMAC formation in T cells and therapeutic perspectives Key words: Immunological Synapses, Supramolecular Activation Cluster, Glioma, viral infection, and immunotherapy Corresponding Author: Carlos Barcia Institute of Neuroscience Department of Biochemistry and Molecular Biology School of Medicine, Lab M2-107 Autonomous University of Barcelona Cerdanyola del Vallès, 08193, Barcelona, Spain [email protected]

Kupfer-type Immunological Synapses in vivo: Raison D’être ......1 Kupfer-type Immunological Synapses in vivo: Raison D’être of SMAC Izaskun Mitxitorena, Elena Saavedra, Carlos

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

  • 1

    Kupfer-type Immunological Synapses in vivo: Raison D’être of SMAC Izaskun Mitxitorena, Elena Saavedra, Carlos Barcia, Ph.D. Department of Biochemistry and Molecular Biology, Institute of Neuroscience & School of Medicine, Universitat Autonoma de Barcelona, Lab M2-107, Bellaterra, Cerdanyola del Valles, Barcelona, Spain. Running Title: SMAC formation in T cells and therapeutic perspectives Key words: Immunological Synapses, Supramolecular Activation Cluster, Glioma, viral infection, and immunotherapy Corresponding Author: Carlos Barcia Institute of Neuroscience

    Department of Biochemistry and Molecular Biology School of Medicine, Lab M2-107 Autonomous University of Barcelona Cerdanyola del Vallès, 08193, Barcelona, Spain [email protected]

  • 2

    Abstract

    T cells engage with antigen-presenting cells to form immunological synapses. These

    intimate contacts are characterized by the complex arrangement of molecules at the

    intercellular interface, which has been described as the supramolecular activation cluster

    (SMAC). However, due to T cells functioning without SMAC formation and the

    difficulties of studying these complex arrangements in vivo, its biological importance has

    been questioned. In light of recent data, we focus this review on the putative functionality

    of SMACs in T-cell synaptic contacts in vivo and emphasize the therapeutic potential of

    SMAC manipulation in immune-driven diseases.

  • 3

    Immunological Synapse Formation and SMAC arrangement

    Immunological synapses (IS) are critical intercellular communications between specific

    immune cells and antigen-presenting cells (APC)1. This particular engagement between

    both counterparts requires intimate contact between the aforementioned cells and

    includes multiple factors and complex signaling cascades of activation 1,2. T-cell ISs have

    been largely studied and represent the best-known IS type 3, although ISs may also be

    established by different types of effector cells, such as NK or B cells 4-6. The formation of

    an IS involves the T-cell recognition of specific antigens that are presented by APCs.

    Major Histocompatibility Complexes (MHC) display antigens at the APC cell surface,

    which are detected by T-cell receptor (TCR) molecules that are displayed on the T-cell

    membrane 7. The interaction between the antigen-MHC and the TCR induces the TCR

    signaling cascade 8, thus initiating the activation of the T cell, which is characterized by

    the phosphorylation and polarization of tyrosine kinases such as lymphocyte-specific

    protein tyrosine kinase (Lck) and zeta-chain-associated protein kinase 70 (ZAP-70) at the

    interface 9,10 (Figure 1). In mature IS formation, the process of activation involves severe

    changes to the micro-anatomical configuration of the T cell that are characterized by

    rearrangement of the actin cytoskeleton and are driven by the microtubules organizer

    center (MTOC), which becomes polarized toward the APC and participates in the

    organization of secretory domains 11-14. The polarization of the T cell is also accompanied

    by the rearrangement of lymphocyte function-associated antigen 1 (LFA-1) molecules

    that segregate three-dimensionally at the IS interface and specifically bind to the APC’s

    intercellular adhesion molecule 1 (ICAM-1) 15,16. This binding of LFA-1/ICAM-1 takes

    place at the interface, and LFA-1/ICAM-1 complexes rearrange micro-anatomically,

  • 4

    forming a ring-shaped area named the peripheral supramolecular activation cluster

    (pSMAC), which surrounds a characteristic central accumulation of TCRs, known as the

    central supramolecular activation cluster (cSMAC) 15 (Figure 1 and Box). This way, a

    “bull’s eye” characteristic structure is formed, where an outer ring contains the adhesion

    molecules, and an inner area contains the signaling molecules. In cytolytic T cells, the

    cSMAC may also contain secretory domains that usually encompass an area of smaller

    size and is located near the TCR signaling central cluster, where lytic granules of effector

    molecules are concentrated and released 6,17,18. Importantly, LFA-1 molecules are linked

    to talin proteins, which are key integrins involved in cell migration and cellular junction

    because they are linked to the actin-myosin cytoskeleton through vinculin 15,19,20.

    Visualization of SMACs in vivo

    The initial description and most of the studies on the microanatomy and function of ISs

    have been performed in vitro 1,15,21. Although the knowledge on ISs has substantially

    grown and successfully improved based on in vitro experiments, the functionality of ISs

    in living organisms has barely been explored. A criticism often rises considering that in

    vitro environments are different from those in tissue. Cultures and planar bilayers are

    isolated, two-dimensional milieus, whereas tissues are three-dimensional environments in

    which cells receive information and signals from different planes and directions

    involving diverse biological systems. Thus, research of ISs in vivo is an important matter

    for a complete understanding of T-cell biology.

    Formation of SMAC in vivo has been demonstrated using high-resolution

    confocal microscopy of labeled, fixed tissue with multiple fluorescence-specific

  • 5

    antibodies. The formation of the CD3/TCR central cluster (cSMAC) and/or the peripheral

    segregation of LFA-1 (pSMAC) are observed in different tissues, such as the brain and

    secondary lymphoid tissues 22,23. ISs are stable and preserved structures in mammals. As

    described in vitro, ISs show a flat interface in vivo; and cSMAC and pSMAC are formed

    in all species studied so far. From rodents 22 to primates 24, including humans 25, the

    formation of SMAC seems to be consistently involved in mammalian immune responses.

    However, despite the good level of resolution, this in vivo technique has the

    limitation of picturing static events. High-resolution confocal images in fixed tissue

    represent a scenery taken at a certain and specific moment and do not resolve the

    dynamics of the IS. Two-photon microscopy in living animals will be the ideal technical

    approach to show the dynamics of IS formation in vivo, but some issues must still be

    solved. Currently, multi-photon microscopes are able to image several hundreds of

    microns deep into tissue; however, the resolution of the anatomical details is still not

    sufficient to distinguish the micro-anatomy of the IS at the SMAC level. In addition,

    observations are hampered by the parenchyma’s high auto-fluorescence and by the

    reduced number of fluorophores that are available to detect molecule arrangements in

    vivo in time-lapse, live imaging. Two-photon microscopy studies in tissue, especially in

    lymph nodes, have shown the dynamics by which T cells engage APCs (i.e., dendritic

    cells), but no micro-anatomical details of the SMAC were given 26,27. Currently, time-

    lapse studies of the microanatomy of complete SMAC formation, containing the central

    and peripheral clusters, have not been yet performed in living tissue. Notably, however, a

    successful attempt was performed regarding visualization of the dynamics of the

    formation of the TCR central cluster using a two-photon microscope in lymph nodes in

  • 6

    live mice. In a study by Friedman et al., some features of the TCR dynamics in vivo, as

    well as the behavior of TCR accumulation, were revealed 28. In addition Azar et al., using

    linker for activation of T cells (LAT)-EGFP labeled T cells, were able to detect the in

    vivo formation of central and peripheral clusters of LAT at the IS interface in lymph

    nodes, which may underlie some insights into the molecular distribution of SMACs 29.

    The next scientific challenge is the combination of different fluorophores to observe the

    dynamics of the peripheral SMAC in relation to the central TCR cluster and how the

    formation of these structures affects immune responses in healthy subjects and

    experimental models of diseases.

    Function of SMAC in vivo

    Previous observations have shown that SMAC formation is not required for TCR

    signaling or for the effectiveness of cytotoxic T cells 6,30. These results question the

    biological importance of SMAC formation. Why is such an enormous and complex

    arrangement in the cell needed? Why invest such a large amount of energy and effort?

    pSMAC and cSMAC formations were first observed in brain tissue, in the context of the

    clearance of virus-infected cells 22. In this case, the formation of SMACs preceded the

    elimination of viral-infected cells in immune-competent animals that were primed with

    an adaptive immune response 22. In this context, the percentage of ISs forming SMACs

    and engaged with virus-infected cells was approximately 60% in a specific time window,

    before complete viral clearance 31,32. These results indicate that a large percentage of

    SMAC formation may be essential for viral clearance in tissue, suggesting its biological

    significance 31. In the same scenario of viral clearance, the secretory domain that was

  • 7

    observed at the immunological synaptic interface was characterized by the formation of

    interferon-gamma (IFN- ) and perforin clusters, which conveys that both effector

    molecules and their polarization at the synaptic interface may be necessary phenomena

    for the elimination of virus-infected cells 31. In fact, IFN- - or perforin-deficient mice are

    unable to eliminate virus-infected cells from the brain 33. However, whether completely

    mature SMAC rearrangements will take place at the interface seems to depend upon

    multiple factors. For example, IFN- appears polarized in Kupfer type (with SMAC) and

    non-Kupfer type (without SMAC) synapses 31, which indicates that the formation of

    mature synapses with SMAC does not precede the formation of the secretory domain;

    therefore, SMAC formation may not be strictly necessary for the release of effector

    molecules and elimination of target cells. In fact, although cytotoxic ISs restrict killing to

    antigenic target cells, IFN- signaling is also detected in non-antigenic bystander cells 34,

    suggesting a certain leakage or multidirectional diffusion of the cytokine, which implies

    defective SMAC formation.

    On the other hand, secretory effector molecules have a different pattern of

    segregation that is independent of c- and pSMAC formation. Therefore, different

    cytokines show different patterns of secretion in T cells. For example, IFN- and

    interleukin 2 (IL-2) are polarized and secreted to the synaptic interface, while TNF- and

    chemokine (C-C motif) ligand 3 (CCL3) are secreted multi-directionally 35. These

    established patterns of secretion indicate a different behavior of T-cells that depends on

    the context of the immune response. Thus, the need for complex SMAC rearrangement

    may not always be required.

  • 8

    These results indicate that SMAC arrangement could be necessary to directionally

    secrete specific molecules towards the APC without altering adjacent cells, thus safely

    channeling intercellular communication 13,36 (Figure 2). Outer ring LFA-1/ICAM-1

    adhesion allows for the formation of a shielded micro-chamber, which is an intercellular

    space that is kept isolated from the surrounding environment. This flat interface feature is

    possible due to rearrangement of the actin cytoskeleton, which forms a consistent and

    renewable scaffold that is oriented to the interface 37,38. Most likely, the reason for these

    interface arrangements may be for maximal reduction of the surface at the intercellular

    contact, which could result in more effective communication and less chance of

    membrane and receptor miss-folding. In that intercellular space, cytotoxic compounds,

    such as effector molecules, can be safely delivered, and signaling only occurs with the

    contacting cell, without damaging the surrounding healthy cells that are not involved in

    the immunological response. Therefore, the formation of SMACs may represent a highly

    evolved and specific immune response that only has an effect on target cells and does not

    affect bystander cells. Thus, it can be hypothesized that the SMAC is a necessary

    structure to channel cytokines and other effector molecules in an extremely selective

    manner (Figure 2).

    Overall, T-cell synaptic contacts may be necessary for an effective immune

    response, but, the formation of SMACs may depend on the immunological context and

    the effector molecules that are delivered. It is, therefore, tempting to speculate that the

    ideal situation may be SMAC formation because it would preserve the surrounding tissue

    and result in a more specific and safe response. As a drawback, an immune response with

    SMAC formation is most likely slower and requires high-energy waste. Thus, if the

  • 9

    immune response needs to be faster and inexpensive, it should be carried out without

    SMAC.

    In summary, in vivo studies of Kupfer-type ISs exhibit a complex scenario for

    further research. Multiple types of intercellular combinations, involving diverse cytokine

    release and adaptable immune responses within different tissues, are important variables

    that should be considered for future research, although the visualization and unraveling of

    the IS function will only be fully achieved in vivo if new, specific approaches are

    designed that selectively inhibit IS formation in the tissue of a living organism.

    A therapeutic view of the immunological synapse

    Because the formation of the SMAC may be an important part of the specificity and

    effectiveness of the T-cell response, manipulation of ISs represents a promising tool from

    a therapeutic point of view. It presents an advantage whereby we could specifically

    inhibit or activate the different immune responses according to therapeutic needs, as

    multiple targets could potentially be aimed to hinder or empower IS formation. In fact,

    immunotherapy is a therapeutic field that has lately been developed and is becoming

    promising, particularly for cancer. Specific drugs, usually artificially made antibodies,

    have been designed to empower anti-tumor immunity, and most of them intervene at the

    synaptic level (Figure 3).

    One of the most hopeful approaches to directly stimulate the formation of specific

    ISs between T and tumor cells is the development of bi-specific T-cell engager (BiTE)

    antibodies. These monoclonal antibodies target the TCR/CD3 complex and tumor

    antigens, such as CD19, epithelial cell adhesion molecule (EpCAM) or epidermal growth

  • 10

    factor receptor (EGFR). This way, the antibodies promote the synaptic interaction

    between tumor cells and T cells and induce the activation of cytolytic T cells. This

    engagement-induced tumor-cell death leads to T-cell accumulation in the tumor

    microenvironment and reduces tumor cell proliferation in vivo 39.

    Another successful approach to modify synaptic contacts is based on the

    development of antibodies that are able to antagonize receptors that inhibit the immune

    response. A successful case is that of ipilimumab, an antibody that binds an inhibitory T-

    cell protein called cytotoxic T lymphocyte antigen 4 (CTLA-4). CTLA-4 is expressed in

    activated T cells and is recruited to the cSMAC in competition with the T-cell activation

    molecule, CD28 40,41. The binding of ipilimumab interferes with CTLA-4-mediated T-cell

    suppression at the cSMAC, therefore, facilitating active synaptic interactions between T

    cells and target cells, which results in a more aggressive immune response against the

    tumor. Ipilimumab has been tested in patients with melanoma (Yervoy®), and it has been

    proven to be effective in specific cases because it removes melanoma without tumor

    recurrence 42-44. Analogously, therapeutic blockade of programmed cell death 1 (PD-1),

    which is also localized at the cSMAC, increases T-cell motility and cytotoxic

    effectiveness, thus improving viral clearance 45. Indeed, the combination of both, CTLA-

    4 and PD-1 blockade, has been proven to be effective toward tumors by increasing the

    cytolytic T-cell population and reducing regulatory T cells 46.

    In this context, optimization of the cytolytic arm seems to be the primary therapeutic

    strategy to eliminate tumors because the tumorigenic microenvironment facilitates a pro-

    inflammatory response that promotes tumor growth. In the case of CNS tumors,

    particularly in human glioma, the formation of SMAC has been studied in depth. In

  • 11

    glioma tissue, mature ISs are established between T-cells and tumorigenic cells, although

    at a low rate 25. However, SMAC analyses performed in murine experimental models of

    glioma have shown that the formation of Kupfer-type synapses does not predict the

    elimination of the tumor 47, which is different from the process of viral clearance 31. This

    feature may be characteristic of tumors because the multidirectional delivery of cytotoxic

    compounds could theoretically be the fastest and most effective way to destroy tumors in

    an environment where the majority of bystander cells should be rapidly eliminated.

    However, because T cells form SMACs, they may still be needed in a sufficient quantity

    for the recognition of specific antigens to take place. This fact supports the idea that

    SMACs would only be formed when the tissue in the vicinity must be preserved. These

    concepts may open new avenues of research regarding the formation of SMAC or

    bonafide ISs.

    On the other hand, tumor development and other immune-mediated degenerative

    diseases might be a consequence of defective SMAC formation. This alteration may be

    reflected in altered immune responses due to deficient recognition of the antigen, anergy

    or exhaustion of the T-cell response, either of the regulatory or cytolytic response. In line

    with this, a recent study showed for the first time that alterations in SMAC formation in T

    cells can be a crucial element in immune disorders. In this report, CD4 T cells obtained

    from patients with multiple sclerosis and type-1 diabetes were exposed to antigens from

    influenza virus. Both CD4-T-cell groups showed divergent formation of SMAC when

    compared with normal T cells obtained from healthy patients 48. These differences

    included deficient SMAC-structure formation regarding the proper CD3/TCR or MHC

    accumulation and ICAM-1/LFA-1 segregation, a distinct motility of T cells, and altered

  • 12

    timing and velocity of SMAC formation. Importantly, a deficiency in SMAC formation

    sets the possibility for alteration in cellular communication and could explain how T cells

    might escape the negative selection that takes place in autoimmune diseases.

    Another example regarding the X-linked lymphoproliferative syndrome, which is

    characterized by fatal responses to Epstein-Barr virus infection, has recently been

    reported. This syndrome is caused by mutations affecting the adaptor SAP (signaling

    lymphocytic activation molecule (SLAM)-associated membrane protein), which is a

    molecule involved in correct arrangement of the synaptic contact. In fact, SAP-deficient

    cytotoxic T lymphocytes exhibit abnormal actin organization and reduced centrosome

    docking at T-cell–B-cell ISs 49. These results demonstrate that correct assembling of T

    cells with their target cells and the micro-anatomical arrangement of SMACs and their

    associated organelles is a fundamental process in the immune response.

    We are beginning to understand how malfunction of SMAC formation may induce

    different immune-mediated diseases. The understanding of this process in vivo as well as

    the specific mechanisms occurring during SMAC formation in tissues within different

    immune scenarios will be crucial to propose molecular targets that restore the correct

    arrangement of Kupfer-type ISs.

    Box

    The term immunological synapse has been used to generally define communications

    between immune cells, although it is also specifically and more accurately referred to as

    the formation of the characteristic interface with complex rearrangements of molecules

    and compounds called SMAC (Supra-Molecular Activation Cluster). Synapses that form

  • 13

    SMACs are considered mature immunological synapses and, in some publications, to

    honor its discoverer, immunological synapses are classified as Kupfer-type or non-

    Kupfer-type immunological synapses according to the presence or absence of the “bull’s

    eye” formation at the interface, respectively.

  • 14

    Figure Legends

    Figure 1. T-cell immunological synapse forming a SMAC (Kupfer-type). T cells

    recognize antigens that are presented by the MHC of an APC through the TCR/CD3

    complex. Then, T cells are activated through phosphorylation of tyrosine kinases such as

    Lck and ZAP-70, which are polarized to the T-cell/APC interface. This activation leads

    to dramatic changes in the cell, including the rearrangement of adhesion molecules, such

    as LFA-1, which are segregated towards the interface to bind ICAM-1 of the APC and

    form the peripheral activation cluster (pSMAC). On the other hand, TCR/CD3 molecules

    are aggregated at the center of the interface and form the central SMAC (cSMAC). In

    addition, cytotoxic granules are delivered to the center of the interface and form the

    secretory domain.

    Figure 2. Hypothetical strategies for cytolytic T-cell responses in tissue. A.

    Unidirectional secretion of effector molecules after immunological synapse formation. T

    cells (red) form mature immunological synapses after antigen recognition and subsequent

    apposition to an APC (blue). LFA-1 adhesion molecules are segregated at the external

    border of the interface (red), forming the pSMAC, whereas TCR (green) is concentrated

    at the center of the interface, forming the cSMAC, where the cytolytic granules (yellow

    arrow) may be delivered in one specific direction. With this strategy, the APC (blue) can

    be specifically eliminated without damaging bystander cells (light brown cells). B.

    Multidirectional secretion of effector molecules without bona fide synapse formation. T

    cells (red) may not form mature immunological synapses after antigen recognition; thus,

  • 15

    the strict apposition to antigen-presenting cells (blue) may not be necessary. LFA-1

    molecules (red) do not arrange as pSMAC, and TCR does not concentrate at the center of

    the interface, forming the cSMAC. Cytolytic granules (yellow arrows) may be delivered

    multi-directionally. With this strategy, bystander APCs (blue) can be eliminated

    discretionally.

    Figure 3. Therapeutic targets at the immunological synapse. CTLA-4 competes with

    CD28 for CD80/CD86. Bound CTLA4-CD80/CD86 complexes are recruited to the

    cSMAC, whereas unbound CD28 is segregated to the pSMAC. PD1 molecules bind to

    PDL1 and are recruited to the cSMAC. The binding of CTLA4-CD80/CD86 inhibits T-

    cell activation. Thus, CTLA-4 blocking antibodies hamper binding to CD80/CD86,

    which facilitates the binding of CD80/CD86 with CD28 and impedes CTL inhibition.

    Similarly, PD1-blocking antibodies obstruct the inhibition of T cells at the synaptic

    interface.

  • 16

    References

    1 Bromley, S. K. et al. The immunological synapse. Annual review of immunology 19, 375-396, doi:10.1146/annurev.immunol.19.1.375 (2001).

    2 Krummel, M. F. & Davis, M. M. Dynamics of the immunological synapse: finding, establishing and solidifying a connection. Current opinion in immunology 14, 66-74 (2002).

    3 Trautmann, A. & Valitutti, S. The diversity of immunological synapses. Current opinion in immunology 15, 249-254 (2003).

    4 Orange, J. S. Formation and function of the lytic NK-cell immunological synapse. Nature reviews. Immunology 8, 713-725, doi:10.1038/nri2381 (2008).

    5 Harwood, N. E. & Batista, F. D. Early events in B cell activation. Annual review of immunology 28, 185-210, doi:10.1146/annurev-immunol-030409-101216 (2010).

    6 Davis, D. & Dustin, M. What is the importance of the immunological synapse? Trends in immunology 25, 323-327, doi:10.1016/j.it.2004.03.007 (2004).

    7 Konig, R. Interactions between MHC molecules and co-receptors of the TCR. Current opinion in immunology 14, 75-83 (2002).

    8 Lee, K. H. et al. T cell receptor signaling precedes immunological synapse formation. Science 295, 1539-1542, doi:10.1126/science.1067710 (2002).

    9 Holdorf, A. D., Lee, K. H., Burack, W. R., Allen, P. M. & Shaw, A. S. Regulation of Lck activity by CD4 and CD28 in the immunological synapse. Nature immunology 3, 259-264, doi:10.1038/ni761 (2002).

    10 Yokosuka, T. et al. Newly generated T cell receptor microclusters initiate and sustain T cell activation by recruitment of Zap70 and SLP-76. Nature immunology 6, 1253-1262, doi:10.1038/ni1272 (2005).

    11 Kuhn, J. R. & Poenie, M. Dynamic polarization of the microtubule cytoskeleton during CTL-mediated killing. Immunity 16, 111-121 (2002).

    12 Stinchcombe, J. C., Bossi, G., Booth, S. & Griffiths, G. M. The immunological synapse of CTL contains a secretory domain and membrane bridges. Immunity 15, 751-761 (2001).

    13 Stinchcombe, J. C., Majorovits, E., Bossi, G., Fuller, S. & Griffiths, G. M. Centrosome polarization delivers secretory granules to the immunological synapse. Nature 443, 462-465, doi:10.1038/nature05071 (2006).

    14 Combs, J. et al. Recruitment of dynein to the Jurkat immunological synapse. Proceedings of the National Academy of Sciences of the United States of America 103, 14883-14888, doi:10.1073/pnas.0600914103 (2006).

    15 Monks, C. R., Freiberg, B. A., Kupfer, H., Sciaky, N. & Kupfer, A. Three-dimensional segregation of supramolecular activation clusters in T cells. Nature 395, 82-86, doi:10.1038/25764 (1998).

    16 Dustin, M. L. et al. A novel adaptor protein orchestrates receptor patterning and cytoskeletal polarity in T-cell contacts. Cell 94, 667-677 (1998).

    17 Irvine, D. J. Function-specific variations in the immunological synapses formed by cytotoxic T cells. Proceedings of the National Academy of Sciences of the United States of America 100, 13739-13740, doi:10.1073/pnas.2536626100 (2003).

  • 17

    18 Lieberman, J. The ABCs of granule-mediated cytotoxicity: new weapons in the arsenal. Nature reviews. Immunology 3, 361-370, doi:10.1038/nri1083 (2003).

    19 Lin, J., Miller, M. J. & Shaw, A. S. The c-SMAC: sorting it all out (or in). The Journal of cell biology 170, 177-182, doi:10.1083/jcb.200503032 (2005).

    20 Critchley, D. R. & Gingras, A. R. Talin at a glance. Journal of cell science 121, 1345-1347, doi:10.1242/jcs.018085 (2008).

    21 Grakoui, A. The Immunological Synapse: A Molecular Machine Controlling T Cell Activation. Science 285, doi:10.1126/science.285.5425.221 (1999).

    22 Barcia, C. et al. In vivo mature immunological synapses forming SMACs mediate clearance of virally infected astrocytes from the brain. The Journal of experimental medicine 203, 2095-2107, doi:10.1084/jem.20060420 (2006).

    23 Khanna, K. M., McNamara, J. T. & Lefrancois, L. In situ imaging of the endogenous CD8 T cell response to infection. Science 318, 116-120, doi:10.1126/science.1146291 (2007).

    24 Barcia, C. et al. CD20, CD3, and CD40 ligand microclusters segregate three-dimensionally in vivo at B-cell-T-cell immunological synapses after viral immunity in primate brain. Journal of virology 82, 9978-9993, doi:10.1128/JVI.01326-08 (2008).

    25 Barcia, C., Jr. et al. Infiltrating CTLs in human glioblastoma establish immunological synapses with tumorigenic cells. The American journal of pathology 175, 786-798, doi:10.2353/ajpath.2009.081034 (2009).

    26 Bousso, P. T-cell activation by dendritic cells in the lymph node: lessons from the movies. Nature reviews. Immunology 8, 675-684, doi:10.1038/nri2379 (2008).

    27 Germain, R. N., Robey, E. A. & Cahalan, M. D. A decade of imaging cellular motility and interaction dynamics in the immune system. Science 336, 1676-1681, doi:10.1126/science.1221063 (2012).

    28 Friedman, R. S., Beemiller, P., Sorensen, C. M., Jacobelli, J. & Krummel, M. F. Real-time analysis of T cell receptors in naive cells in vitro and in vivo reveals flexibility in synapse and signaling dynamics. The Journal of experimental medicine 207, 2733-2749, doi:10.1084/jem.20091201 (2010).

    29 Azar, G. A., Lemaitre, F., Robey, E. A. & Bousso, P. Subcellular dynamics of T cell immunological synapses and kinapses in lymph nodes. Proceedings of the National Academy of Sciences of the United States of America 107, 3675-3680, doi:10.1073/pnas.0905901107 (2010).

    30 Purbhoo, M. A., Irvine, D. J., Huppa, J. B. & Davis, M. M. T cell killing does not require the formation of a stable mature immunological synapse. Nature immunology 5, 524-530, doi:10.1038/ni1058 (2004).

    31 Barcia, C. et al. In vivo polarization of IFN-gamma at Kupfer and non-Kupfer immunological synapses during the clearance of virally infected brain cells. J Immunol 180, 1344-1352 (2008).

    32 Barcia, C. et al. T cells' immunological synapses induce polarization of brain astrocytes in vivo and in vitro: a novel astrocyte response mechanism to cellular injury. PloS one 3, e2977, doi:10.1371/journal.pone.0002977 (2008).

    33 Zirger, J. M. et al. Immune-mediated Loss of Transgene Expression From Virally Transduced Brain Cells Is Irreversible, Mediated by IFNgamma, Perforin, and TNFalpha, and due to the Elimination of Transduced Cells. Molecular therapy :

  • 18

    the journal of the American Society of Gene Therapy 20, 808-819, doi:10.1038/mt.2011.243 (2012).

    34 Sanderson, N. S. et al. Cytotoxic immunological synapses do not restrict the action of interferon-gamma to antigenic target cells. Proceedings of the National Academy of Sciences of the United States of America 109, 7835-7840, doi:10.1073/pnas.1116058109 (2012).

    35 Huse, M., Lillemeier, B. F., Kuhns, M. S., Chen, D. S. & Davis, M. M. T cells use two directionally distinct pathways for cytokine secretion. Nature immunology 7, 247-255 (2006).

    36 Dustin, M. L., Chakraborty, A. K. & Shaw, A. S. Understanding the Structure and Function of the Immunological Synapse. Cold Spring Harbor Perspectives in Biology 2, doi:10.1101/cshperspect.a002311 (2010).

    37 Dustin, M. L. & Cooper, J. A. The immunological synapse and the actin cytoskeleton: molecular hardware for T cell signaling. Nature immunology 1, 23-29, doi:10.1038/76877 (2000).

    38 Vicente-Manzanares, M. & Sanchez-Madrid, F. Role of the cytoskeleton during leukocyte responses. Nature reviews. Immunology 4, 110-122, doi:10.1038/nri1268 (2004).

    39 Lutterbuese, R. et al. T cell-engaging BiTE antibodies specific for EGFR potently eliminate KRAS- and BRAF-mutated colorectal cancer cells. Proceedings of the National Academy of Sciences of the United States of America 107, 12605-12610, doi:10.1073/pnas.1000976107 (2010).

    40 Walker, L. S. & Sansom, D. M. The emerging role of CTLA4 as a cell-extrinsic regulator of T cell responses. Nature reviews. Immunology 11, 852-863, doi:10.1038/nri3108 (2011).

    41 Yokosuka, T. et al. Spatiotemporal basis of CTLA-4 costimulatory molecule-mediated negative regulation of T cell activation. Immunity 33, 326-339, doi:10.1016/j.immuni.2010.09.006 (2010).

    42 Lipson, E. J. & Drake, C. G. Ipilimumab: an anti-CTLA-4 antibody for metastatic melanoma. Clinical cancer research : an official journal of the American Association for Cancer Research 17, 6958-6962, doi:10.1158/1078-0432.CCR-11-1595 (2011).

    43 Hodi, F. S. et al. Improved survival with ipilimumab in patients with metastatic melanoma. The New England journal of medicine 363, 711-723, doi:10.1056/NEJMoa1003466 (2010).

    44 Pardoll, D. M. Immunology beats cancer: a blueprint for successful translation. Nature immunology 13, 1129-1132, doi:10.1038/ni.2392 (2012).

    45 Zinselmeyer, B. H. et al. PD-1 promotes immune exhaustion by inducing antiviral T cell motility paralysis. The Journal of experimental medicine 210, 757-774, doi:10.1084/jem.20121416 (2013).

    46 Curran, M. A., Montalvo, W., Yagita, H. & Allison, J. P. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proceedings of the National Academy of Sciences of the United States of America 107, 4275-4280, doi:10.1073/pnas.0915174107 (2010).

  • 19

    47 Yang, J. et al. Kupfer-type immunological synapse characteristics do not predict anti-brain tumor cytolytic T-cell function in vivo. Proceedings of the National Academy of Sciences of the United States of America 107, 4716-4721, doi:10.1073/pnas.0911587107 (2010).

    48 Schubert, D. A. et al. Self-reactive human CD4 T cell clones form unusual immunological synapses. The Journal of experimental medicine 209, 335-352, doi:10.1084/jem.20111485 (2012).

    49 Zhao, F., Cannons, J. L., Dutta, M., Griffiths, G. M. & Schwartzberg, P. L. Positive and negative signaling through SLAM receptors regulate synapse organization and thresholds of cytolysis. Immunity 36, 1003-1016, doi:10.1016/j.immuni.2012.05.017 (2012).

  • Article FileFigure 1Figure 2Figure 3

    Texto1: Post-print of: "Kupfer-type immunological synapses in vivo: Raison D’être of SMAC / I. Mitxitorena, E. Saavedra and C. Barcia", in Immunology and Cell Biology (Nature), 2015, vol. 93, p. 51–56; doi:10.1038/icb.2014.80